Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Drug Metab Dispos ; 49(12): 1047-1055, 2021 12.
Article in English | MEDLINE | ID: mdl-34593616

ABSTRACT

Exemestane (EXE) is a hormonal therapy used to treat estrogen receptor-positive breast cancer by inhibiting the final step of estrogen biosynthesis catalyzed by the enzyme aromatase. Cysteine conjugates of EXE and its active metabolite 17ß-dihydro-EXE (DHE) are the major metabolites found in both the urine and plasma of patients taking EXE. The initial step in cysteine conjugate formation is glutathione conjugation catalyzed by the glutathione S-transferase (GST) family of enzymes. The goal of the present study was to identify cytosolic hepatic GSTs active in the GST-mediated metabolism of EXE and 17ß-DHE. Twelve recombinant cytosolic hepatic GSTs were screened for their activity against EXE and 17ß-DHE, and glutathionylated EXE and 17ß-DHE conjugates were detected by ultra-performance liquid chromatography tandem mass spectrometry. GST α (GSTA) isoform 1, GST µ (GSTM) isoform 3 and isoform 1 were active against EXE, whereas only GSTA1 exhibited activity against 17ß-DHE. GSTM1 exhibited the highest affinity against EXE with a Michaelis-Menten constant (KM) value that was 3.8- and 7.1-fold lower than that observed for GSTA1 and GSTM3, respectively. Of the three GSTs, GSTM3 exhibited the highest intrinsic clearance against EXE (intrinsic clearance = 0.14 nl·min-1·mg-1). The KM values observed for human liver cytosol against EXE (46 µM) and 17ß-DHE (77 µM) were similar to those observed for recombinant GSTA1 (53 and 30 µM, respectively). Western blot analysis revealed that GSTA1 and GSTM1 composed 4.3% and 0.57%, respectively, of total protein in human liver cytosol; GSTM3 was not detected. These data suggest that GSTA1 is the major hepatic cytosolic enzyme involved in the clearance of EXE and its major active metabolite, 17ß-DHE. SIGNIFICANCE STATEMENT: Most previous studies related to the metabolism of the aromatase inhibitor exemestane (EXE) have focused mainly on phase I metabolic pathways and the glucuronidation phase II metabolic pathway. However, recent studies have indicated that glutathionylation is the major metabolic pathway for EXE. The present study is the first to characterize hepatic glutathione S-transferase (GST) activity against EXE and 17ß-dihydro-EXE and to identify GST α 1 and GST µ 1 as the major cytosolic GSTs involved in the hepatic metabolism of EXE.


Subject(s)
Androstadienes/pharmacokinetics , Breast Neoplasms , Glutathione Transferase/metabolism , Inactivation, Metabolic/physiology , Liver/enzymology , Antineoplastic Agents, Hormonal/pharmacokinetics , Aromatase Inhibitors/pharmacokinetics , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Chromatography, Liquid , Cysteine/metabolism , Cytosol/metabolism , Estrogens/biosynthesis , Glutathione Transferase/chemistry , Hepatobiliary Elimination/physiology , Humans , Protein Isoforms , Receptors, Estrogen
2.
Drug Metab Dispos ; 46(12): 1867-1878, 2018 12.
Article in English | MEDLINE | ID: mdl-30257855

ABSTRACT

Exemestane (EXE) is an aromatase inhibitor used for the prevention and treatment of estrogen receptor-positive breast cancer. Although the known major metabolic pathway for EXE is reduction to form the active 17ß-dihydro-EXE (17ß-DHE) and subsequent glucuronidation to 17ß-hydroxy-EXE-17-O-ß-D-glucuronide (17ß-DHE-Gluc), previous studies have suggested that other major metabolites exist for exemestane. In the present study, a liquid chromatography-mass spectrometry (LC-MS) approach was used to acquire accurate mass data in MSE mode, in which precursor ion and fragment ion data were obtained simultaneously to screen novel phase II EXE metabolites in urine specimens from women taking EXE. Two major metabolites predicted to be cysteine conjugates of EXE and 17ß-DHE by elemental composition were identified. The structures of the two metabolites were confirmed to be 6-methylcysteinylandrosta-1,4-diene-3,17-dione (6-EXE-cys) and 6-methylcysteinylandrosta-1,4-diene-17ß-hydroxy-3-one (6-17ß-DHE-cys) after comparison with their chemically synthesized counterparts. Both underwent biosynthesis in vitro in three stepwise enzymatic reactions, with the first involving glutathione conjugation. The cysteine conjugates of EXE and 17ß-DHE were subsequently quantified by liquid chromatography-mass spectrometry in the urine and matched plasma samples of 132 subjects taking EXE. The combined 6-EXE-cys plus 6-17ß-DHE-cys made up 77% of total EXE metabolites in urine (vs. 1.7%, 0.14%, and 21% for EXE, 17ß-DHE, and 17ß-DHE-Gluc, respectively) and 35% in plasma (vs. 17%, 12%, and 36% for EXE, 17ß-DHE, and 17ß-DHE-Gluc, respectively). Therefore, cysteine conjugates of EXE and 17ß-DHE appear to be major metabolites of EXE in both urine and plasma.


Subject(s)
Androstadienes/metabolism , Aromatase Inhibitors/metabolism , Breast Neoplasms , Adult , Aged , Aged, 80 and over , Androstadienes/administration & dosage , Androstadienes/blood , Androstadienes/urine , Aromatase Inhibitors/administration & dosage , Aromatase Inhibitors/blood , Aromatase Inhibitors/urine , Breast Neoplasms/blood , Breast Neoplasms/urine , Chromatography, Liquid , Cysteine/metabolism , Female , Glucuronides/metabolism , Humans , Metabolic Detoxication, Phase II , Middle Aged , Tandem Mass Spectrometry
3.
J Med Chem ; 61(16): 7065-7086, 2018 08 23.
Article in English | MEDLINE | ID: mdl-29995408

ABSTRACT

Cigarette smoking causes nearly one in every five deaths in the United States. The development of a specific inhibitor of cytochrome P450 2A6 (CYP2A6), the major nicotine-metabolizing enzyme in humans, which could be prescribed for the cessation of cigarette smoking, has been undertaken. To further refine the structure activity relationship of CYP2A6, previously synthesized 3-alkynyl and 3-heteroaromatic substituted pyridine methanamines were used as lead compounds. Isosteric pyridine replacement and appendage of all available positions around the pyridine ring with a methyl group was performed to identify a modification that would increase CYP2A6 inhibition potency, which led to 4g (IC50 = 0.055 mM) as a primary analogue. Potent compounds were evaluated for CYP selectivity, human liver microsomal half-life, and compliance with the rules of five. Top compounds (i.e., 6i, IC50 = 0.017 mM, >120 min half-life) are poised for further development as treatments for smoking and tobacco use cessation.


Subject(s)
Cytochrome P-450 CYP2A6/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Methylamines/pharmacology , Microsomes, Liver/drug effects , Pyridines/pharmacology , Cytochrome P-450 CYP2A6/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Methylamines/chemical synthesis , Methylamines/chemistry , Microsomes, Liver/enzymology , Molecular Structure , Pyridines/chemical synthesis , Pyridines/chemistry , Smoking/drug therapy , Tobacco Use Cessation
4.
Pharmacol Res Perspect ; 5(3): e00313, 2017 06.
Article in English | MEDLINE | ID: mdl-28603632

ABSTRACT

Exemestane (EXE) treats estrogen receptor positive (ER+) breast cancer in postmenopausal women by inhibiting the estrogen-synthesizing cytochrome P450 CYP19A1. Variability in the severity and incidence of side effects as well as overall drug efficacy may be partially explained by genetic factors, including nonsynonymous variation in CYP19A1, also known as aromatase. The present study identified phase I EXE metabolites in human liver microsomes (HLM) and investigated mechanisms that may alter the extent of systemic estrogen deprivation in EXE-treated women with breast cancer, including whether functional polymorphisms in aromatase cause differential inhibition by EXE and whether EXE metabolites possess anti-aromatase activity. The potency of EXE and ten of its derivatives was measured with HEK293-overexpressed wild type aromatase (CYP19A1*1) using a rapid novel UPLC tandem mass spectrometry method. Of the ten compounds assayed, five were poor inhibitors (IC 50 Ëƒ 50 µmol/L) of wild type aromatase while five others, including the major metabolite, 17ß-dihydroexemestane (17ß-DHE), exhibited moderate potency, with IC 50 values ranging between 1.2 and 7.1 µmol/L. The anti-aromatase activity of EXE was also tested with two common allozymes, aromataseThr201Met (CYP19A1*3) and aromataseArg264Cys (CYP19A1*4). Differential inhibition of variant aromatase is unlikely to account for variable clinical outcomes as EXE-mediated inhibition of aromataseThr201Met (IC 50 = 0.86 ± 0.12 µmol/L) and aromataseArg264Cys (IC 50 = 1.7 ± 0.65 µmol/L) did not significantly differ from wild type (IC 50 = 0.92 ± 0.17 µmol/L). Although less potent than the parent drug, these results suggest that active metabolites may contribute to the therapeutic mechanism of EXE.

5.
Pharmacol Res Perspect ; 5(3): e00314, 2017 06.
Article in English | MEDLINE | ID: mdl-28603633

ABSTRACT

Exemestane (EXE) is an endocrine therapy commonly used by postmenopausal women with hormone-responsive breast cancer due to its potency in inhibiting aromatase-catalyzed estrogen synthesis. Preliminary in vitro studies sought to identify phase I EXE metabolites and hepatic cytochrome P450s (CYP450s) that participate in EXE biotransformation. Phase I metabolites were identified by incubating EXE with HEK293-overexpressed CYP450s. CYP450s 1A2, 2C8, 2C9, 2C19, 2D6, 3A4, and 3A5 produce 17ß-dihydroexemestane (17ß-DHE), an active major metabolite, as well as two inactive metabolites. 17ß-DHE formation in pooled human liver microsomes subjected to isoform-specific CYP450 inhibition was also monitored using tandem mass spectrometry. 17ß-DHE production in human liver microsomes was unaffected by isoform-specific inhibition of CYP450s 2A6, 2B6, and 2E1 but decreased 12-39% following inhibition of drug-metabolizing enzymes from CYP450 subfamilies 1A, 2C, 2D, and 3A. These results suggest that redundancy exists in the EXE metabolic pathway with multiple hepatic CYP450s catalyzing 17ß-DHE formation in vitro. To further expand the knowledge of phase I EXE metabolism, the impact of CYP450 genetic variation on 17ß-DHE formation was assessed via enzyme kinetic parameters. Affinity for EXE substrate and enzyme catalytic velocity were calculated for hepatic wild-type CYP450s and their common nonsynonymous variants by monitoring the reduction of EXE to 17ß-DHE. Several functional polymorphisms in xenobiotic-metabolizing CYP450s 1A2, 2C8, 2C9, and 2D6 resulted in deviant enzymatic activity relative to wild-type enzyme. Thus, it is possible that functional polymorphisms in EXE-metabolizing CYP450s contribute to inter-individual variability in patient outcomes by mediating overall exposure to the drug and its active metabolite, 17ß-DHE.

6.
Pharmacogenet Genomics ; 26(8): 370-80, 2016 08.
Article in English | MEDLINE | ID: mdl-27111237

ABSTRACT

OBJECTIVE: Exemestane (EXE) is a potent third-generation aromatase inhibitor used as endocrine therapy in breast cancer treatment and prevention. Characterization of its metabolic pathway is incomplete, with ambiguity existing in the identity of enzymes driving the production of its key metabolite, 17ß-dihydroexemestane (17ß-DHE). The impact of genetic variation on EXE metabolism is also unknown. This study aims to describe cytosolic reductase involvement in hepatic EXE metabolism and to assess the impact of functional polymorphisms on metabolite production. MATERIALS AND METHODS: Phase I metabolites were identified in incubations of EXE with pooled human liver cytosol or recombinant protein for AKR1Cs and CBR1. Kinetic parameters characterizing EXE reduction were measured for purified wild-type enzymes, and nonsynonymous variants occurring at greater than 1% minor allele frequency using UPLC/MS/MS. RESULTS: Human liver cytosol, CBR1, AKR1C1, AKR1C2, AKR1C3, and AKR1C4 reduce EXE to active primary metabolite 17ß-DHE. The formation of a novel metabolite, 17α-DHE, was catalyzed by recombinant AKR1C4 and CBR1 in addition to hepatic cytosol. Variants AKR1C3 Arg258Cys and AKR1C4 Gly135Glu had significantly decreased affinity for EXE relative to their respective wild types. Five common AKR1C3 polymorphisms were associated with decreased rates of catalysis, whereas AKR1C4 Gly135Glu increased the velocity of EXE reduction. CONCLUSION: AKR1Cs and CBR1 catalyze EXE reduction in vitro. These results imply that cytosolic ketosteroid reductases may participate in the EXE metabolic pathway in vivo. In addition, several common variants were associated with altered enzymatic activity, suggesting that functional polymorphisms could play an important role in overall EXE metabolism and activity by altering the extent and duration of 17ß-DHE exposure.


Subject(s)
Androstadienes/metabolism , Cytosol/enzymology , Liver/enzymology , Oxidoreductases/genetics , Polymorphism, Single Nucleotide/genetics , Catalysis , Humans , In Vitro Techniques , Kinetics , Metabolomics , Tandem Mass Spectrometry
7.
Curr Drug Deliv ; 10(5): 557-63, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23755721

ABSTRACT

3'-O-Retinoyl-5-fluoro-2'-deoxyuridine (RFUdR) is a putative dual-acting, mutually-masking (DAMM) prodrug for the treatment of cancer. As part of the proof of principle for the DAMM concept, the concentrations of RFUdR and its post-hydrolysis active metabolites, 5-fluoro-2'-deoxyuridine (FUdR) and all-trans-retinoic acid (RA), were determined in plasma and selected tissues following either bolus intravenous (i.v.; 12.5 µmol/kg) or oral (p.o.; 13.7 µmol/kg) doses of RFUdR to mice bearing EMT6 murine mammary tumors. The concentrations of RFUdR and its primary metabolites were measured by high-performance liquid chromatography. A three compartment model provided the best fit for plasma RFUdR after an i.v. bolus, whereas FUdR and RA data were best fit by a one compartment model. The terminal half-life of RFUdR in plasma was 9 hours. The AUC of RFUdR in tumor (3400 µmol/L.min) was estimated to be about 4- fold higher than its AUC in the plasma (809 ± 241 µmol/L.min). A short-duration, saturated elimination phase for RFUdR was observed in both liver and kidney following an i.v. bolus. Neither unchanged RFUdR nor RA was detected in urine. The high bioavailability (~90%) following oral dosing with RFUdR indicates that this DAMM prodrug may be suitable for oral dosing to deliver FUdR and RA for cancer chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Floxuridine/analogs & derivatives , Floxuridine/pharmacokinetics , Neoplasms/metabolism , Prodrugs/pharmacokinetics , Animals , Antineoplastic Agents/blood , Cell Line, Tumor , Female , Floxuridine/blood , Kidney/metabolism , Liver/metabolism , Lung/metabolism , Mice , Mice, Inbred BALB C
8.
Mol Cancer Ther ; 10(11): 2052-61, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21885864

ABSTRACT

The ceramide/sphingosine-1-phosphate (S1P) rheostat has been hypothesized to play a critical role in regulating tumor cell fate, with elevated levels of ceramide inducing death and elevated levels of S1P leading to survival and proliferation. Ceramidases are key enzymes that control this rheostat by hydrolyzing ceramide to produce sphingosine and may also confer resistance to drugs and radiation. Therefore, ceramidase inhibitors have excellent potential for development as new anticancer drugs. In this study, we identify a novel ceramidase inhibitor (Ceranib-1) by screening a small molecule library and describe the synthesis of a more potent analogue (Ceranib-2). In a cell-based assay, both compounds were found to inhibit cellular ceramidase activity toward an exogenous ceramide analogue, induce the accumulation of multiple ceramide species, decrease levels of sphingosine and S1P, inhibit the proliferation of cells alone and in combination with paclitaxel, and induce cell-cycle arrest and cell death. In vivo, Ceranib-2 was found to delay tumor growth in a syngeneic tumor model without hematologic suppression or overt signs of toxicity. These data support the selection of ceramidases as suitable targets for anticancer drug development and provide the first nonlipid inhibitors of human ceramidase activity.


Subject(s)
Antineoplastic Agents/pharmacology , Ceramidases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Quinolones/pharmacology , Adenocarcinoma/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Enzyme Inhibitors/therapeutic use , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mice , Mice, Inbred BALB C , Quinolones/therapeutic use , Small Molecule Libraries
9.
Proc Natl Acad Sci U S A ; 108(2): 528-33, 2011 Jan 11.
Article in English | MEDLINE | ID: mdl-21187426

ABSTRACT

The serine/threonine Pim kinases are overexpressed in solid cancers and hematologic malignancies and promote cell growth and survival. Here, we find that a novel Pim kinase inhibitor, SMI-4a, or Pim-1 siRNA blocked the rapamycin-sensitive mammalian target of rapamycin (mTORC1) activity by stimulating the phosphorylation and thus activating the mTORC1 negative regulator AMP-dependent protein kinase (AMPK). Mouse embryonic fibroblasts (MEFs) deficient for all three Pim kinases [triple knockout (TKO) MEFs] demonstrated activated AMPK driven by elevated ratios of AMPATP relative to wild-type MEFs. Consistent with these findings, TKO MEFs were found to grow slowly in culture and have decreased rates of protein synthesis secondary to a diminished amount of 5'-cap-dependent translation. Pim-3 expression alone in TKO MEFs was sufficient to reverse AMPK activation, increase protein synthesis, and drive MEF growth similar to wild type. Pim-3 expression was found to markedly increase the protein levels of both c-Myc and the peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α), enzymes capable of regulating glycolysis and mitochondrial biogenesis, which were diminished in TKO MEFs. Overexpression of PGC-1α in TKO MEFs elevated ATP levels and inhibited the activation of AMPK. These results demonstrate the Pim kinase-mediated control of energy metabolism and thus regulation of AMPK activity. We identify an important role for Pim-3 in modulating c-Myc and PGC-1α protein levels and cell growth.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Proteins/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Trans-Activators/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival , Fibroblasts/metabolism , Heat-Shock Proteins/metabolism , Humans , K562 Cells , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Proto-Oncogene Proteins c-myc/metabolism , TOR Serine-Threonine Kinases , Transcription Factors/metabolism
10.
Bioorg Med Chem ; 18(3): 1003-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20085856

ABSTRACT

Substantial interest has focused on the roles of sphingolipid metabolizing enzymes in a variety of hyperproliferative and inflammatory diseases. A key family of enzymes involved in these pathologies is the ceramidases. Ceramidases cleave the pro-apoptotic lipid ceramide into a long-chain fatty acid and sphingosine, which can then be further metabolized to the mitogenic and inflammatory lipid sphingosine 1-phosphate. Consequently, development of ceramidase inhibitors would provide useful pharmacologic probes for further studies of sphingolipid metabolism, as well as lead compounds for drug development. This effort has been hampered by the lack of in vitro and cellular ceramidase assays that are amenable to high-throughput screening. Recently, a fluorogenic ceramide analog has been described as a substrate for use in ceramidase assays. The synthesis of this compound has now been substantially improved in terms of both the required effort and the overall yield of the process. Key improvements include: reduction in number of required steps, use of a hydroboration reaction; incorporation of a Mitsunobu reaction; improved acylation by the addition of triethylamine; together providing a fourfold increase in the overall yield. In addition, it has been demonstrated that the ceramide analog can be used in high-throughput assays to identify ceramidase inhibitors. Overall, the improved efficiency in the preparation of this ceramidase substrate should accelerate discovery efforts relating to sphingolipid metabolism.


Subject(s)
Ceramidases/metabolism , Ceramides/chemical synthesis , Fluorescent Dyes/chemical synthesis , Cell Line , Ceramidases/antagonists & inhibitors , Ceramides/chemistry , Fluorescent Dyes/chemistry , Humans , Molecular Structure
11.
Blood ; 115(4): 824-33, 2010 Jan 28.
Article in English | MEDLINE | ID: mdl-19965690

ABSTRACT

The serine/threonine Pim kinases are up-regulated in specific hematologic neoplasms, and play an important role in key signal transduction pathways, including those regulated by MYC, MYCN, FLT3-ITD, BCR-ABL, HOXA9, and EWS fusions. We demonstrate that SMI-4a, a novel benzylidene-thiazolidine-2, 4-dione small molecule inhibitor of the Pim kinases, kills a wide range of both myeloid and lymphoid cell lines with precursor T-cell lymphoblastic leukemia/lymphoma (pre-T-LBL/T-ALL) being highly sensitive. Incubation of pre-T-LBL cells with SMI-4a induced G1 phase cell-cycle arrest secondary to a dose-dependent induction of p27(Kip1), apoptosis through the mitochondrial pathway, and inhibition of the mammalian target of rapamycin C1 (mTORC1) pathway based on decreases in phospho-p70 S6K and phospho-4E-BP1, 2 substrates of this enzyme. In addition, treatment of these cells with SMI-4a was found to induce phosphorylation of extracellular signal-related kinase1/2 (ERK1/2), and the combination of SMI-4a and a mitogen-activated protein kinase kinase 1/2 (MEK1/2) inhibitor was highly synergistic in killing pre-T-LBL cells. In immunodeficient mice carrying subcutaneous pre-T-LBL tumors, treatment twice daily with SMI-4a caused a significant delay in the tumor growth without any change in the weight, blood counts, or chemistries. Our data suggest that inhibition of the Pim protein kinases may be developed as a therapeutic strategy for the treatment of pre-T-LBL.


Subject(s)
Benzylidene Compounds/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Thiazolidinediones/pharmacology , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , G1 Phase/drug effects , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Jurkat Cells , MAP Kinase Signaling System/drug effects , Mice , Mice, Knockout , Mice, Transgenic , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , TOR Serine-Threonine Kinases
12.
Mol Cancer Ther ; 8(6): 1473-83, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19509254

ABSTRACT

The Pim protein kinases play important roles in cancer development and progression, including prostate tumors and hematologic malignancies. To investigate the potential role of these enzymes as anticancer drug targets, we have synthesized novel benzylidene-thiazolidine-2,4-diones that function as potent Pim protein kinase inhibitors. With IC(50) values in the nanomolar range, these compounds block the ability of Pim to phosphorylate peptides and proteins in vitro and, when added to DU145 prostate cancer cells overexpressing Pim, inhibit the ability of this enzyme to phosphorylate a known substrate, the BH(3) protein BAD. When added to prostate cancer cell lines, including PC3, DU145, and CWR22Rv1, and human leukemic cells, MV4;11, K562, and U937 cells, these compounds induce G(1)-S cell cycle arrest and block the antiapoptotic effect of the Pim protein kinase. The cell cycle arrest induced by these compounds is associated with an inhibition of cyclin-dependent kinase 2 and activity and translocation of the Pim-1 substrate p27(Kip1), a cyclin-dependent kinase 2 inhibitory protein, to the nucleus. Furthermore, when added to leukemic cells, these compounds synergize with the mammalian target of rapamycin inhibitor rapamycin to decrease the phosphorylation level of the translational repressor 4E-BP1 at sites phosphorylated by mammalian target of rapamycin. Combinations of rapamycin and the benzylidene-thiazolidine-2,4-diones synergistically block the growth of leukemic cells. Thus, these agents represent novel Pim inhibitors and point to an important role for the Pim protein kinases in cell cycle control in multiple types of cancer cells.


Subject(s)
Benzylidene Compounds/pharmacology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Thiazolidinediones/pharmacology , Apoptosis/drug effects , Benzylidene Compounds/chemistry , Blotting, Western , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Intracellular Signaling Peptides and Proteins/metabolism , K562 Cells , Leukemia/metabolism , Leukemia/pathology , Male , Molecular Structure , Phosphorylation/drug effects , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-pim-1/metabolism , Thiazolidinediones/chemistry , U937 Cells , bcl-Associated Death Protein/metabolism
13.
J Med Chem ; 52(1): 74-86, 2009 Jan 08.
Article in English | MEDLINE | ID: mdl-19072652

ABSTRACT

The Pim protein kinases are frequently overexpressed in prostate cancer and certain forms of leukemia and lymphoma. 5-(3-Trifluoromethylbenzylidene)thiazolidine-2,4-dione (4a) was identified by screening to be a Pim-1 inhibitor and was found to attenuate the autophosphorylation of tagged Pim-1 in intact cells. Although 4a is a competitive inhibitor with respect to ATP, a screen of approximately 50 diverse protein kinases demonstrated that it has high selectivity for Pim kinases. Computational docking of 4a to Pim-1 provided a model for lead optimization, and a series of substituted thiazolidine-2,4-dione congeners was synthesized. The most potent new compounds exhibited IC(50)s of 13 nM for Pim-1 and 2.3 microM for Pim-2. Additional compounds in the series demonstrated selectivities of more than 2500-fold and 400-fold for Pim-1 or Pim-2, respectively, while other congeners were essentially equally potent toward the two isozymes. Overall, these compounds are new Pim kinase inhibitors that may provide leads to novel anticancer agents.


Subject(s)
Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Thiazolidinediones/chemical synthesis , Thiazolidinediones/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line , Cell-Free System , Combinatorial Chemistry Techniques , Female , Humans , Kinetics , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Structure , Neoplasm Transplantation , Neoplasms/drug therapy , Neoplasms/pathology , Phosphorylation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-pim-1/metabolism , Structure-Activity Relationship , Thiazolidinediones/chemistry , Thiazolidinediones/therapeutic use
14.
Dig Dis Sci ; 53(4): 997-1012, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18058233

ABSTRACT

A critical step in the mechanism of action of inflammatory cytokines is the stimulation of sphingolipid metabolism, including activation of sphingosine kinase (SK), which produces the mitogenic and proinflammatory lipid sphingosine 1-phosphate (S1P). We have developed orally bioavailable compounds that effectively inhibit SK activity in vitro in intact cells and in cancer models in vivo. In this study, we assessed the effects of these SK inhibitors on cellular responses to tumor necrosis factor alpha (TNFalpha) and evaluated their efficacy in the dextran sulfate sodium (DSS) model of ulcerative colitis in mice. Using several cell systems, it was shown that the SK inhibitors block the ability of TNFalpha to activate nuclear factor kappa B (NFkappaB), induce expression of adhesion proteins, and promote production of prostaglandin E(2) (PGE(2)). In an acute model of DSS-induced ulcerative colitis, SK inhibitors were equivalent to or more effective than Dipentum in reducing disease progression, colon shortening, and neutrophil infiltration into the colon. The effects of SK inhibitors were associated with decreased colonic levels of inflammatory cytokines TNFalpha, interleukin (IL)-1beta, interferon gamma (IFN)-gamma, IL-6, and reduction of S1P levels. A similar reduction in disease progression was provided by SK inhibitors in a chronic model of ulcerative colitis in which the mice received 3-week-long cycles of DSS interspaced with week-long recovery periods. In the chronic model, immunohistochemistry for SK showed increased expression in DSS-treated mice (compared with water-treated controls) that was reduced by drug treatment. S1P levels were also elevated in the DSS group and significantly reduced by drug treatment. Together, these data indicate that SK is a critical component in inflammation and that inhibitors of this enzyme may be useful in treating inflammatory bowel diseases.


Subject(s)
Colitis, Ulcerative/drug therapy , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Administration, Oral , Animals , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Cytokines/metabolism , Dextran Sulfate , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphotransferases (Alcohol Group Acceptor)/administration & dosage , Tumor Necrosis Factor-alpha/metabolism
15.
J Lipid Res ; 48(8): 1873-84, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17525474

ABSTRACT

Many important signaling proteins require the posttranslational addition of fatty acid chains for their proper subcellular localization and function. One such modification is the addition of palmitoyl moieties by enzymes known as palmitoyl acyltransferases (PATs). Substrates for PATs include C-terminally farnesylated proteins, such as H- and N-Ras, as well as N-terminally myristoylated proteins, such as many Src-related tyrosine kinases. The molecular and biochemical characterization of PATs has been hindered by difficulties in developing effective methods for the analysis of PAT activity. In this study, we describe the use of cell-permeable, fluorescently labeled lipidated peptides that mimic the PAT recognition domains of farnesylated and myristoylated proteins. These PAT substrate mimetics are accumulated by SKOV3 cells in a saturable and time-dependent manner. Although both peptides are rapidly palmitoylated, the SKOV3 cells have a greater capacity to palmitoylate the myristoylated peptide than the farnesylated peptide. Confocal microscopy indicated that the palmitoylated peptides colocalized with Golgi and plasma membrane markers, whereas the corresponding nonpalmitoylatable peptides accumulated in the Golgi but did not traffic to the plasma membrane. Overall, these studies indicate that the lipidated peptides provide useful cellular probes for quantitative and compartmentalization studies of protein palmitoylation in intact cells.


Subject(s)
Acyltransferases/metabolism , Lipoproteins/metabolism , Palmitic Acid/metabolism , Peptides/chemistry , Peptides/metabolism , Female , Humans , Kinetics , Lipoproteins/chemistry , Microscopy, Confocal , Protein Transport , Time Factors , Tumor Cells, Cultured
16.
Methods ; 40(2): 166-70, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17012028

ABSTRACT

Protein palmitoylation is emerging as an important post-translational modification in development as well as in the establishment and progression of diseases such as cancer. This chapter describes the use of fluorescent lipidated peptides to characterize palmitoyl acyltransferase (PAT) activities in vitro and in intact cells. The peptides mimic two motifs that are enzymatically palmitoylated, i.e. C-terminal farnesyl and N-terminal myristoyl sequences. These substrate peptides can be separated from the palmitoylated product peptides by reversed-phase HPLC, detected and quantified by the fluorescence of their NBD label. Through these methods, the activities of PATs toward these alternate substrates in isolated membranes or intact cells can be quantified. The in vitro assay has been used to characterize human PATs and to identify inhibitors of these enzymes. The cellular assay has been useful in elucidating the kinetics of protein palmitoylation by PATs in situ, and the sub-cellular distribution of the palmitoylated products.


Subject(s)
Acyltransferases/analysis , Fluorescent Dyes , Lipoproteins , Palmitic Acid/metabolism , Peptides
17.
Mol Cancer Ther ; 5(7): 1647-59, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16891450

ABSTRACT

The covalent attachment of palmitate to specific proteins by the action of palmitoyl acyltransferases (PAT) plays critical roles in the biological activities of several oncoproteins. Two PAT activities are expressed by human cells: type 1 PATs that modify the farnesyl-dependent palmitoylation motif found in H- and N-Ras, and type 2 PATs that modify the myristoyl-dependent palmitoylation motif found in the Src family of tyrosine kinases. We have previously shown that the type 1 PAT HIP14 causes cellular transformation. In the current study, we show that mRNA encoding HIP14 is up-regulated in a number of types of human tumors. To assess the potential of HIP14 and other PATs as targets for new anticancer drugs, we developed three cell-based assays suitable for high-throughput screening to identify inhibitors of these enzymes. Using these screens, five chemotypes, with activity toward either type 1 or type 2 PAT activity, were identified. The activity of the hits were confirmed using assays that quantify the in vitro inhibition of PAT activity, as well as a cell-based assay that determines the abilities of the compounds to prevent the localization of palmitoylated green fluorescent proteins to the plasma membrane. Representative compounds from each chemotype showed broad antiproliferative activity toward a panel of human tumor cell lines and inhibited the growth of tumors in vivo. Together, these data show that PATs, and HIP14 in particular, are interesting new targets for anticancer compounds, and that small molecules with such activity can be identified by high-throughput screening.


Subject(s)
Acyltransferases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Carrier Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Neoplasms/enzymology , Nerve Tissue Proteins/antagonists & inhibitors , Acyltransferases/genetics , Adaptor Proteins, Signal Transducing , Antineoplastic Agents/chemistry , Carrier Proteins/genetics , Cell Line, Tumor , Drug Screening Assays, Antitumor/methods , Drug Screening Assays, Antitumor/statistics & numerical data , Enzyme Inhibitors/chemistry , Humans , Neoplasms/genetics , Nerve Tissue Proteins/genetics , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , ras Proteins/antagonists & inhibitors , ras Proteins/metabolism
18.
J Med Chem ; 47(6): 1413-22, 2004 Mar 11.
Article in English | MEDLINE | ID: mdl-14998330

ABSTRACT

In a search for improved multiple drug resistance (MDR) modulators, we identified a novel series of substituted pyrroloquinolines that selectively inhibits the function of P-glycoprotein (Pgp) without modulating multidrug resistance-related protein 1 (MRP1). These compounds were evaluated for their toxicity toward drug-sensitive tumor cells (i.e. MCF-7, T24) and for their ability to antagonize Pgp-mediated drug-resistant cells (i.e. NCI/ADR) and MRP1-mediated resistant cells (i.e. MCF-7/VP). Cytotoxicity and drug accumulation assays demonstrated that the dihydropyrroloquinolines inhibit Pgp to varying degrees, without any significant inhibition of MRP1. The compound termed PGP-4008 was the most effective at inhibiting Pgp in vitro and was further evaluated in vivo. PGP-4008 inhibited tumor growth in a murine syngeneic Pgp-mediated MDR solid tumor model when given in combination with doxorubicin. PGP-4008 was rapidly absorbed after intraperitoneal administration, with its plasma concentrations exceeding the in vitro effective dose for more than 2 h. PGP-4008 did not alter the plasma distribution of concomitantly administered anticancer drugs and did not cause systemic toxicity as was observed for cyclosporin A. Because of their enhanced selectivity toward Pgp, these substituted dihydropyrroloquinolines may be effective MDR modulators in a clinical setting.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Acetanilides , Antineoplastic Agents/chemical synthesis , Drug Resistance, Multiple , Pyrroles/chemical synthesis , Quinolines/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Area Under Curve , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Half-Life , Humans , Mice , Mice, Inbred BALB C , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Neoplasm Transplantation , Pyrroles/chemistry , Pyrroles/pharmacology , Quinolines/chemistry , Quinolines/pharmacology , Transplantation, Heterologous
19.
J Pharmacol Exp Ther ; 309(1): 340-7, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14724220

ABSTRACT

N-myristoyltransferase (NMT) is an emerging therapeutic target that catalyzes the attachment of myristate to the N terminus of an acceptor protein. We have developed a medium-throughput assay for screening potential small molecule inhibitors of human NMT-1 consisting of recombinant enzyme, biotinylated peptide substrate, and [3H]myristoyl-CoA. Approximately 16,000 diverse compounds have been evaluated, and significant inhibition of NMT was found with 0.8% of the compounds. From these hits, we have identified the cyclohexyl-octahydropyrrolo[1,2-a]pyrazine (COPP) chemotype as inhibitory toward human NMT-1. Thirty-two compounds containing this substructure inhibited NMT-1, with IC(50) values ranging from 6 microM to millimolar concentrations, and a quantitative structure-activity relationship equation (r(2) = 0.72) was derived for the series. The most potent inhibitor (24, containing 9-ethyl-9H-carbazole) demonstrated competitive inhibition for the peptide-binding site of NMT-1 and noncompetitive inhibition for the myristoyl-CoA site. Computational docking studies using the crystal structure of the highly homologous yeast NMT confirmed that 24 binds with excellent complementarity to the peptide-binding site of the enzyme. To evaluate the ability of 24 to inhibit NMT activity in intact cells, monkey CV-1 cells expressing an N-myristoylated green fluorescent protein (GFP) fusion protein were treated with a known NMT inhibitor or with 24. Each compound caused the redistribution of GFP from the plasma membrane to the cytosol. Furthermore, 24 inhibits cancer cell proliferation at doses similar to those that inhibit protein myristoylation. Overall, these studies establish an efficient assay for screening for inhibitors of human NMT and identify a novel family of inhibitors that compete at the peptide-binding site and have activity in intact cells.


Subject(s)
Acyltransferases/metabolism , Enzyme Inhibitors/pharmacology , Pyrazines/pharmacology , Pyrroles/pharmacology , Cell Division/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Pyrazines/chemical synthesis , Pyrazines/chemistry , Pyrroles/chemical synthesis , Pyrroles/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
20.
J Inorg Biochem ; 98(2): 287-300, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14729309

ABSTRACT

The coenzymic activity of eight analogs of coenzyme B(12) (5'-deoxyadenosyl-cobalamin, AdoCbl) with structural alterations in the Ado ligand has been investigated with the AdoCbl-dependent ribonucleoside triphosphate reductase (RTPR) from Lactobacillus leichmannii. Six of the analogs were partially active coenzymes, and one, 3-iso-5'-deoxyadenosylcobalamin (3-IsoAdoCbl) was nearly as active as AdoCbl itself. NMR-restrained molecular modeling of 3-IsoAdoCbl revealed a highly conformationally mobile structure which required a four state model to be consistent with the NMR data. Thus, two conformations, one with the IsoAdo ligand over the eastern quadrant of the corrin, and one with the IsoAdo ligand over the northern quadrant, each undergo a facile syn/anti conformational equilibrium in the IsoAdo ligand. Spectrophotometric measurement of the kinetics of RTPR-induced cleavage of the carbon-cobalt bond of 3-IsoAdoCbl showed that it binds to the enzyme with the same affinity as AdoCbl, but its homolysis is only 20% as rapid. Investigation of the non-enzymatic thermolysis of 3-IsoAdoCbl showed that like AdoCbl, 3-IsoAdoCbl decomposes by competing homolytic and heterolytic pathways. A complete temperature-dependent kinetic and product analysis, followed by correction for the base-off species permitted deconvolution of the specific rate constant for both pathways. Eyring plots for the homolysis and heterolysis rate constant cross at 93 degrees C, so that homolysis is the predominant pathway at high temperature, but heterolysis is the predominant pathway at low temperature. At 37 degrees C, the homolysis of 3-IsoAdoCbl is 5.5-fold faster than that of AdoCbl, and the enzyme catalyzes carbon-cobalt bond homolysis in 3-IsoAdoCbl by a factor of 5.9 x 10(7), only 3.9% of the catalytic efficiency with AdoCbl itself. It seems likely that the conformational flexibility of 3-IsoAdoCbl allows it to adopt a coformation in which the hydrogen bonding patterns of the adenine moiety are similar to those of AdoCbl itself, and that this is responsible for the high enzymatic activity of this analog.


Subject(s)
Cobamides/chemistry , Cobamides/metabolism , Carbon/chemistry , Cobalt/chemistry , Hydrogen Bonding , Kinetics , Propanediol Dehydratase/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Solutions
SELECTION OF CITATIONS
SEARCH DETAIL
...